EV-D68

EV - D68
  • 文章类型: Comparative Study
    焦亡,促炎的程序性细胞死亡,与2019年冠状病毒病和其他病毒性疾病的发病机理有关。Gasdermin家族蛋白(GSDMs),包括GSDMD和GSDME,是焦转细胞死亡的关键调节因子。然而,病毒感染调节焦亡的机制尚不清楚.这里,我们采用mCherry-GSDMD荧光报告基因检测方法来筛选阻碍GSDMD在活细胞中定位和功能的病毒蛋白.我们的数据表明,严重急性呼吸综合征冠状病毒2(SARS-CoV-2)的主要蛋白酶NSP5通过切割GSDMD的残基Q29和Q193阻断了GSDMD介导的焦亡。而另一种SARS-CoV-2蛋白酶,NSP3在残基G370处切割GSDME,但激活GSDME介导的焦亡。有趣的是,呼吸道肠道病毒EV-D68编码的蛋白酶3C和2A也通过灭活GSDMD但启动GSDME介导的焦亡而对GSDM的功能表现出相似的差异调节。EV-D68感染通过诱导细胞凋亡对人癌细胞产生溶瘤作用。我们的发现提供了有关呼吸道病毒如何操纵宿主细胞焦亡的见解,并为抗病毒治疗和癌症治疗提供了潜在的靶标。IMPORTANCEPyroptosis在2019冠状病毒病的发病机理中起着至关重要的作用,理解其功能可能有助于开发新的治疗策略。本研究旨在探讨病毒编码的蛋白酶如何调节焦亡。我们研究了严重急性呼吸综合征冠状病毒2(SARS-CoV-2)和呼吸道肠道病毒D68(EV-D68)蛋白酶对宿主细胞焦亡的影响。我们发现SARS-CoV-2编码的蛋白酶NSP5和NSP3使gasderminD(GSDMD)失活,但引发gasderminE(GSDME)介导的焦亡,分别。我们还发现另一种呼吸道病毒EV-D68编码两种不同的蛋白酶2A和3C,它们选择性触发GSDME介导的焦亡,同时抑制GSDMD的功能。基于这些发现,我们进一步注意到,EV-D68感染引发人癌细胞的焦亡并产生溶瘤作用.我们的研究为病毒调节的焦亡的分子机制提供了新的见解,并确定了抗病毒和癌症疗法发展的潜在靶标。
    Pyroptosis, a pro-inflammatory programmed cell death, has been implicated in the pathogenesis of coronavirus disease 2019 and other viral diseases. Gasdermin family proteins (GSDMs), including GSDMD and GSDME, are key regulators of pyroptotic cell death. However, the mechanisms by which virus infection modulates pyroptosis remain unclear. Here, we employed a mCherry-GSDMD fluorescent reporter assay to screen for viral proteins that impede the localization and function of GSDMD in living cells. Our data indicated that the main protease NSP5 of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) blocked GSDMD-mediated pyroptosis via cleaving residues Q29 and Q193 of GSDMD. While another SARS-CoV-2 protease, NSP3, cleaved GSDME at residue G370 but activated GSDME-mediated pyroptosis. Interestingly, respiratory enterovirus EV-D68-encoded proteases 3C and 2A also exhibit similar differential regulation on the functions of GSDMs by inactivating GSDMD but initiating GSDME-mediated pyroptosis. EV-D68 infection exerted oncolytic effects on human cancer cells by inducing pyroptotic cell death. Our findings provide insights into how respiratory viruses manipulate host cell pyroptosis and suggest potential targets for antiviral therapy as well as cancer treatment.IMPORTANCEPyroptosis plays a crucial role in the pathogenesis of coronavirus disease 2019, and comprehending its function may facilitate the development of novel therapeutic strategies. This study aims to explore how viral-encoded proteases modulate pyroptosis. We investigated the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and respiratory enterovirus D68 (EV-D68) proteases on host cell pyroptosis. We found that SARS-CoV-2-encoded proteases NSP5 and NSP3 inactivate gasdermin D (GSDMD) but initiate gasdermin E (GSDME)-mediated pyroptosis, respectively. We also discovered that another respiratory virus EV-D68 encodes two distinct proteases 2A and 3C that selectively trigger GSDME-mediated pyroptosis while suppressing the function of GSDMD. Based on these findings, we further noted that EV-D68 infection triggers pyroptosis and produces oncolytic effects in human carcinoma cells. Our study provides new insights into the molecular mechanisms underlying virus-modulated pyroptosis and identifies potential targets for the development of antiviral and cancer therapeutics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    在成功控制脊髓灰质炎病毒后,呼吸道肠道病毒D68(EV-D68)的重新出现,一种突出的非脊髓灰质炎肠道病毒,已成为全球严重的公共卫生问题。宿主先天性免疫反应是抵御EV-D68入侵的主要防御;然而,病毒逃避干扰素(IFN)抗病毒活性的潜在机制尚不清楚。在这项研究中,我们发现EV-D68通过切割信号转导和转录激活因子1(STAT1)抑制I型IFN信号,细胞对干扰素和其他细胞因子反应的关键因素。我们观察到原型和循环EV-D68菌株保留了它们诱导STAT1裂解和减弱IFN信号转导的能力。进一步的研究显示,EV-D683C蛋白酶在131Q残基处切割STAT1。有趣的是,并非所有肠道病毒编码的3C蛋白酶都表现出这种能力。EV-D68和脊髓灰质炎病毒3C蛋白酶有效诱导STAT1裂解;然而,来自EV-A71,柯萨奇病毒A16和回声病毒的3C蛋白酶没有。STAT1裂解还消除了STAT1响应于IFN刺激激活下游信号元件的核易位能力。总的来说,这些结果表明,由病毒蛋白酶3C靶向的STAT1,EV-D68利用它来颠覆宿主的先天免疫反应。IMPORTANCE肠病毒D68(EV-D68)在过去十年中发生了重大变化,从一种罕见的病原体演变为一种潜在的大流行病原体。干扰素(IFN)信号通路是宿主抵抗病毒侵袭的重要防御机制和治疗靶点。以前的研究报道,EV-D68病毒通过不同的策略阻断或削弱宿主细胞中的免疫识别和IFN的产生;然而,EV-D68对IFN信号传导的抗性机制尚未完全阐明。我们的研究表明,EV-D68依赖于自己编码的蛋白酶,3C,直接切割信号转导和转录激活因子1(STAT1),IFN信号通路中的关键转导成分,破坏IFN介导的抗病毒反应。以前对人类肠道病毒的研究没有记载直接裂解STAT1蛋白以逃避细胞免疫防御。然而,并非所有的肠道病毒3C蛋白都能裂解STAT1。这些发现强调了不同人类肠道病毒用来逃避宿主免疫的不同进化策略。
    Following the successful control of poliovirus, the re-emergence of respiratory enterovirus D68 (EV-D68), a prominent non-polio enterovirus, has become a serious public health concern worldwide. Host innate immune responses are the primary defense against EV-D68 invasion; however, the mechanism underlying viral evasion of the antiviral activity of interferons (IFN) remains unclear. In this study, we found that EV-D68 inhibited type I IFN signaling by cleaving signal transducer and activator of transcription 1 (STAT1), a crucial factor in cellular responses to interferons and other cytokines. We observed that the prototype and circulating EV-D68 strains conserved their ability to induce STAT1 cleavage and attenuate IFN signal transduction. Further investigation revealed that EV-D68 3C protease cleaves STAT1 at the 131Q residue. Interestingly, not all enterovirus-encoded 3C proteases exhibited this ability. EV-D68 and poliovirus 3C proteases efficiently induced STAT1 cleavage; whereas, 3C proteases from EV-A71, coxsackievirus A16, and echoviruses did not. STAT1 cleavage also abolished the nuclear translocation capacity of STAT1 in response to IFN stimulation to activate downstream signaling elements. Overall, these results suggest that STAT1, targeted by viral protease 3C, is utilized by EV-D68 to subvert the host\'s innate immune response.IMPORTANCEEnterovirus D68 (EV-D68) has significantly transformed over the past decade, evolving from a rare pathogen to a potential pandemic pathogen. The interferon (IFN) signaling pathway is an important defense mechanism and therapeutic target for the host to resist viral invasion. Previous studies have reported that the EV-D68 virus blocks or weakens immune recognition and IFN production in host cells through diverse strategies; however, the mechanisms of EV-D68 resistance to IFN signaling have not been fully elucidated. Our study revealed that EV-D68 relies on its own encoded protease, 3C, to directly cleave signal transducer and activator of transcription 1 (STAT1), a pivotal transduction component in the IFN signaling pathway, disrupting the IFN-mediated antiviral response. Previous studies on human enteroviruses have not documented direct cleavage of the STAT1 protein to evade cellular immune defenses. However, not all enteroviral 3C proteins can cleave STAT1. These findings highlight the diverse evolutionary strategies different human enteroviruses employ to evade host immunity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肠道病毒D68(EV-D68)是小病毒科肠道病毒属中肠道病毒D种的成员。作为一种新兴的非脊髓灰质炎肠道病毒,EV-D68在世界各地广泛传播,会导致严重的神经系统和呼吸系统疾病。尽管细胞中的内在限制因素提供了前线防御,病毒-宿主相互作用的分子性质仍然难以捉摸。这里,我们提供的证据表明,主要的组织相容性复合体II类伴侣,CD74,通过与2B蛋白的第二个疏水区相互作用抑制EV-D68在感染细胞中的复制,而EV-D68通过3Cpro裂解减弱CD74的抗病毒作用。3Cpro在Gln-125处切割CD74。CD74和EV-D683Cpro之间的平衡决定了病毒感染的结果。重要性作为一种新兴的非脊髓灰质炎肠道病毒,EV-D68在世界各地广泛传播,会导致严重的神经系统和呼吸系统疾病。这里,我们报道,CD74通过靶向EV-D68的2B蛋白抑制感染细胞中的病毒复制,而EV-D68通过3Cpro裂解减弱CD74的抗病毒作用.CD74和EV-D683Cpro之间的平衡决定了病毒感染的结果。
    Enterovirus D68 (EV-D68) is a member of the species Enterovirus D in the genus Enterovirus of the family Picornaviridae. As an emerging non-polio enterovirus, EV-D68 is widely spread all over the world and causes severe neurological and respiratory illnesses. Although the intrinsic restriction factors in the cell provide a frontline defense, the molecular nature of virus-host interactions remains elusive. Here, we provide evidence that the major histocompatibility complex class II chaperone, CD74, inhibits EV-D68 replication in infected cells by interacting with the second hydrophobic region of 2B protein, while EV-D68 attenuates the antiviral role of CD74 through 3Cpro cleavage. 3Cpro cleaves CD74 at Gln-125. The equilibrium between CD74 and EV-D68 3Cpro determines the outcome of viral infection. IMPORTANCE As an emerging non-polio enterovirus, EV-D68 is widely spread all over the world and causes severe neurological and respiratory illnesses. Here, we report that CD74 inhibits viral replication in infected cells by targeting 2B protein of EV-D68, while EV-D68 attenuates the antiviral role of CD74 through 3Cpro cleavage. The equilibrium between CD74 and EV-D68 3Cpro determines the outcome of viral infection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肠道病毒D68(EV-D68)是一种全球新兴的病原体,主要在儿童中引起严重的呼吸道疾病。来自EV-D68的蛋白酶可损害I型干扰素(IFN-I)的产生。然而,EV-D68结构蛋白在拮抗宿主抗病毒反应中的作用仍不清楚.我们发现EV-D68结构蛋白VP3与IFN调节因子7(IRF7)相互作用,这种相互作用抑制了IRF7的磷酸化和核易位,然后抑制了IFN的转录。此外,VP3通过与IRF7的竞争性相互作用抑制TNF受体相关因子6(TRAF6)诱导的IRF7泛素化。IRF7Δ305-503与VP3的相互作用能力较弱,而VP3Δ41-50与IRF7的相互作用能力较弱。还发现来自肠道病毒A71(EV-A71)和柯萨奇病毒A16(CV-A16)的VP3与IRF7蛋白相互作用。这些结果表明,肠道病毒结构蛋白VP3在颠覆宿主先天免疫反应中起着关键作用,可能是抗病毒药物研究的潜在靶标。重要性EV-D68是一种全球新兴的病原体,可导致严重的呼吸道疾病。这里,我们报道EV-D68通过靶向IRF7抑制先天免疫应答.进一步的研究表明,结构蛋白VP3通过与IRF7的竞争性相互作用抑制了TRAF6诱导的IRF7的泛素化。这些结果表明VP3对IRF7的控制可能是EV-D68抑制IFN-I产生的机制。
    Enterovirus D68 (EV-D68) is a globally emerging pathogen causing severe respiratory illnesses mainly in children. The protease from EV-D68 could impair type I interferon (IFN-I) production. However, the role of the EV-D68 structural protein in antagonizing host antiviral responses remains largely unknown. We showed that the EV-D68 structural protein VP3 interacted with IFN regulatory factor 7 (IRF7), and this interaction suppressed the phosphorylation and nuclear translocation of IRF7 and then repressed the transcription of IFN. Furthermore, VP3 inhibited the TNF receptor associated factor 6 (TRAF6)-induced ubiquitination of IRF7 by competitive interaction with IRF7. IRF7Δ305-503 showed much weaker interaction ability to VP3, and VP3Δ41-50 performed weaker interaction ability with IRF7. The VP3 from enterovirus A71 (EV-A71) and coxsackievirus A16 (CV-A16) was also found to interact with the IRF7 protein. These results indicate that the enterovirus structural protein VP3 plays a pivotal role in subverting host innate immune responses and may be a potential target for antiviral drug research. IMPORTANCE EV-D68 is a globally emerging pathogen that causes severe respiratory illnesses. Here, we report that EV-D68 inhibits innate immune responses by targeting IRF7. Further investigations revealed that the structural protein VP3 inhibited the TRAF6-induced ubiquitination of IRF7 by competitive interaction with IRF7. These results indicate that the control of IRF7 by VP3 may be a mechanism by which EV-D68 represses IFN-I production.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肠道病毒D68(EV-D68),导致严重的呼吸系统疾病和不可逆的中枢神经系统损伤,已成为世界性的严重公共卫生问题。然而,EV-D68发挥神经毒性的机制尚不清楚.因此,我们旨在分析EV-D68感染对卵裂的影响,亚细胞易位,和TARDNA结合蛋白43kDa(TDP-43)在呼吸或神经细胞中的致病性聚集。结果表明,EV-D68编码的蛋白酶2A和3C诱导了TDP-43的易位和切割,分别。具体来说,在图3C中,TDP-43的裂解残基327Q。与野生型TDP-43相比,3C介导的切割的TDP-43片段具有显著降低的蛋白质溶解度。因此,3C活动促进了TDP-43的聚集,对不同的人类细胞产生细胞毒性,包括胶质母细胞瘤T98G细胞。筛选了市售抗病毒药物对3C介导的TDP-43裂解的影响,结果表明,洛匹那韦是EV-D683C蛋白酶的有效抑制剂。总的来说,这些结果表明TDP-43是EV-D683C的保守宿主靶标。这项研究首次提供了TDP-43失调参与EV-D68发病机制的证据。重要性在过去的十年里,肠道病毒D68(EV-D68)感染的发病率在全球范围内有所增加.EV-D68感染可引起不同的呼吸道症状和严重的神经系统并发症,包括急性弛缓性脊髓炎.因此,阐明EV-D68毒性的潜在机制对于开发预防EV-D68感染相关疾病的新方法很重要。这项研究表明,EV-D68感染引发了转位,乳沟,和TDP-43的聚集,TDP-43是一种与退行性神经系统疾病密切相关的细胞内蛋白。病毒蛋白酶3C降低了TDP-43的溶解度,从而对宿主细胞施加细胞毒性,包括人类胶质母细胞瘤细胞.因此,抵消3C活性是缓解EV-D68触发的细胞死亡的有效策略。TDP-43的细胞质聚集是退行性疾病的标志,有助于神经细胞损伤和中枢神经系统(CNS)疾病。这项关于EV-D68诱导的TDP-43形成的研究结果扩展了我们对病毒介导的细胞毒性以及感染患者中TDP-43功能障碍相关的认知障碍和神经症状的潜在风险的理解。
    Enterovirus D68 (EV-D68), which causes severe respiratory diseases and irreversible central nervous system damage, has become a serious public health problem worldwide. However, the mechanisms by which EV-D68 exerts neurotoxicity remain unclear. Thus, we aimed to analyze the effects of EV-D68 infection on the cleavage, subcellular translocation, and pathogenic aggregation of TAR DNA-binding protein 43 kDa (TDP-43) in respiratory or neural cells. The results showed that EV-D68-encoded proteases 2A and 3C induced TDP-43 translocation and cleavage, respectively. Specifically, 3C cleaved residue 327Q of TDP-43. The 3C-mediated cleaved TDP-43 fragments had substantially decreased protein solubility compared with the wild-type TDP-43. Hence, 3C activity promoted TDP-43 aggregation, which exerted cytotoxicity to diverse human cells, including glioblastoma T98G cells. The effects of commercially available antiviral drugs on 3C-mediated TDP-43 cleavage were screened, and the results revealed lopinavir as a potent inhibitor of EV-D68 3C protease. Overall, these results suggested TDP-43 as a conserved host target of EV-D68 3C. This study is the first to provide evidence on the involvement of TDP-43 dysregulation in EV-D68 pathogenesis. IMPORTANCE Over the past decade, the incidence of enterovirus D68 (EV-D68) infection has increased worldwide. EV-D68 infection can cause different respiratory symptoms and severe neurological complications, including acute flaccid myelitis. Thus, elucidating the mechanisms underlying EV-D68 toxicity is important to develop novel methods to prevent EV-D68 infection-associated diseases. This study shows that EV-D68 infection triggers the translocalization, cleavage, and aggregation of TDP-43, an intracellular protein closely related to degenerative neurological disorders. The viral protease 3C decreased TDP-43 solubility, thereby exerting cytotoxicity to host cells, including human glioblastoma cells. Thus, counteracting 3C activity is an effective strategy to relieve EV-D68-triggered cell death. Cytoplasmic aggregation of TDP-43 is a hallmark of degenerative diseases, contributing to neural cell damage and central nervous system (CNS) disorders. The findings of this study on EV-D68-induced TDP-43 formation extend our understanding of virus-mediated cytotoxicity and the potential risks of TDP-43 dysfunction-related cognitive impairment and neurological symptoms in infected patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肠道病毒D68(EV-D68)是一种全球重新出现的呼吸道病原体,与严重呼吸道疾病的爆发有关,并与急性弛缓性脊髓炎有关。然而,EV-D68感染的有效疫苗或治疗仍然缺乏。我们证明了蓝莓的活性成分,蝶芪(Pte),以及它的主要代谢产物,pinostilbene(Pin),促进EV-D68感染的人呼吸道细胞的先天免疫反应。Pte和Pin治疗可明显缓解EV-D68引发的细胞病变效应。重要的是,Pte和Pin都破坏了病毒RNA复制(EC50等级从1.336到4.997µM),并以剂量依赖的方式产生了感染性病毒粒子,在杀病毒浓度下没有细胞毒性。Pte或Pin处理的呼吸道细胞对EV-D68进入没有任何影响,但显示出病毒RNA复制和蛋白质合成显着降低。最后,我们表明,Pte和Pin广泛抑制了从最近的大流行中分离出的循环EV-D68菌株的复制能力。总之,我们的结果表明,Pte及其衍生物,销,增强宿主对EV-D68的免疫识别并抑制EV-D68的复制,这代表了抗病毒药物开发的有希望的策略。
    Enterovirus D68 (EV-D68) is a globally re-emerging respiratory pathogen implicated in outbreaks of severe respiratory illnesses and associated with acute flaccid myelitis. However, effective vaccines or treatments for EV-D68 infections remain scarce. We demonstrated that the active constituent of blueberries, pterostilbene (Pte), and its major metabolite, pinostilbene (Pin), facilitated innate immune responses in EV-D68-infected human respiratory cells. Pte and Pin treatment clearly relieved EV-D68-triggered cytopathic effects. Importantly, both Pte and Pin disrupted viral RNA replication (EC50 rank from 1.336 to 4.997 µM) and infectious virion production in a dose-dependent manner, without cytotoxicity at virucidal concentrations. Pte- or Pin-treated respiratory cells did not show any influences on EV-D68 entry but showed substantially decreased viral RNA replication and protein synthesis. Finally, we showed that Pte and Pin broadly suppressed the replication capacity of circulating EV-D68 strains isolated from recent pandemics. In summary, our results suggest that Pte and its derivative, Pin, enhance host immune recognition of EV-D68 and suppress EV-D68 replication, which represents a promising strategy for antiviral drug development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:由人肠道病毒D68(EV-D68)引起的严重呼吸道和神经系统疾病对公众健康构成严重威胁,目前还没有有效的药物和疫苗。腺苷脱氨酶作用于RNA1(ADAR1)在各种病毒感染中具有不同的生物学功能,但其在EV-D68感染中的作用仍未确定.
    方法:横纹肌肉瘤(RD)和人胚肾293T(293T)细胞,和HeLa细胞用于评估EV-D68(费蒙毒株)和人3型副流感病毒(HPIV3;NIH47885)感染后ADAR1的表达水平,分别。通过沉默RNA(siRNA)的敲低和细胞中ADAR1p110或ADAR1p150的过表达用于确定病毒感染后两种蛋白质的功能。使用无缝克隆试剂盒产生ADAR1p110双链RNA结合结构域(dsRBD)缺失突变。使用蛋白质印迹法鉴定ADAR1,EV-D68VP1和HPIV3血凝素-神经氨酸酶(HN)蛋白的表达。应用中位组织培养感染剂量(TCID50)来检测病毒滴度。使用逆转录定量PCR(RT-qPCR)分析EV-D68mRNA的转录水平,并且使用双荧光素酶报告系统分析病毒5'-非翻译区(5'-UTR)介导的翻译。
    结论:我们发现EV-D68感染后ADAR1的转录和表达受到抑制。RNA干扰内源性ADAR1降低VP1蛋白表达和病毒滴度,而过表达ADAR1p110,而不是ADAR1p150,促进病毒复制。免疫荧光检测显示,EV-D68感染后,ADAR1p110从细胞核迁移到胞浆。Further,ADAR1p110在脱氨酶结构域的活性位点突变后失去了其前病毒能力,病毒基因组的5'-UTR测序显示ADAR1p110可能在EV-D68RNA编辑中起作用。此外,在ADAR1击倒后,磷酸化双链RNA依赖性蛋白激酶(p-PKR)和磷酸化真核起始因子2α(p-eIF2α)的水平均增加。在双荧光素酶报告基因测定中还观察到病毒基因组5'-UTR的减弱的翻译活性。最后,ADAR1p110dsRBD的缺失增加了p-PKR的水平,与VP1表达降低相关,表明ADAR1p110促进EV-D68复制也与其dsRBD抑制PKR活化有关。我们的研究表明,ADAR1p110是EV-D68复制的新型病毒原因子,为EV-D68抗病毒研究提供了理论依据。
    Severe respiratory and neurological diseases caused by human enterovirus D68 (EV-D68) pose a serious threat to public health, and there are currently no effective drugs and vaccines. Adenosine deaminase acting on RNA1 (ADAR1) has diverse biological functions in various viral infections, but its role in EV-D68 infections remains undetermined.
    Rhabdomyosarcoma (RD) and human embryonic kidney 293 T (293 T) cells, and HeLa cells were used to evaluate the expression level of ADAR1 upon EV-D68 (Fermon strain) and human parainfluenza virus type 3 (HPIV3; NIH47885) infection, respectively. Knockdown through silencing RNA (siRNA) and overexpression of either ADAR1p110 or ADAR1p150 in cells were used to determine the function of the two proteins after viral infection. ADAR1p110 double-stranded RNA binding domains (dsRBDs) deletion mutation was generated using a seamless clone kit. The expression of ADAR1, EV-D68 VP1, and HPIV3 hemagglutinin-neuraminidase (HN) proteins was identified using western blotting. The median tissue culture infectious dose (TCID50) was applied to detect viral titers. The transcription level of EV-D68 mRNA was analyzed using reverse transcription-quantitative PCR (RT-qPCR) and the viral 5\'-untranslated region (5\'-UTR)-mediated translation was analyzed using a dual luciferase reporter system.
    We found that the transcription and expression of ADAR1 was inhibited upon EV-D68 infection. RNA interference of endogenous ADAR1 decreased VP1 protein expression and viral titers, while overexpression of ADAR1p110, but not ADAR1p150, facilitated viral replication. Immunofluorescence assays showed that ADAR1p110 migrated from the nucleus to the cytoplasm after EV-D68 infection. Further, ADAR1p110 lost its pro-viral ability after mutations of the active sites in the deaminase domain, and 5\'-UTR sequencing of the viral genome revealed that ADAR1p110 likely plays a role in EV-D68 RNA editing. In addition, after ADAR1 knockdown, the levels of both phosphorylated double-stranded RNA dependent protein kinase (p-PKR) and phosphorylated eukaryotic initiation factor 2α (p-eIF2α) increased. Attenuated translation activity of the viral genome 5\'-UTR was also observed in the dual-luciferase reporter assay. Lastly, the deletion of ADAR1p110 dsRBDs increased the level of p-PKR, which correlated with a decreased VP1 expression, indicating that the promotion of EV-D68 replication by ADAR1p110 is also related to the inhibition of PKR activation by its dsRBDs. Our study illustrates that ADAR1p110 is a novel pro-viral factor of EV-D68 replication and provides a theoretical basis for EV-D68 antiviral research.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Long interspersed element 1 (LINE-1 or L1) is the only active autonomous retrotransposon in the human genome that can serve as an endogenous upstream activator of cytoplasmic nucleic acid sensing pathways to elicit an antiviral immune response. In this study, we investigated the influence of enteroviral infection on L1 mobility. The results showed that infection with different enteroviruses, both EV-D68 and EV-A71, blocked L1 transposition. We screened diverse viral accessory proteins for L1 activity and identified EV-D68 2A, 3A, 3C, and EV-A71 ORF2p proteins as viral L1 inhibitors. EV-D68 2A suppressed L1 mobility by expression suppression of L1 proteins. Viral proteins 3A and 3C restricted ORF2p-mediated L1 reverse transcription in isolated L1 ribonucleoproteins. The newly identified enteroviral protein ORF2p inhibited the expression of L1 ORF1p. Altogether, our findings shed light on the strict modulation of L1 retrotransposons during enterovirus replication.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    肠道病毒D68(EV-D68)是一种呼吸道病毒病原体,可引起严重的呼吸道疾病和神经系统表现。自2014年疫情爆发以来,据报道,EV-D68在全球范围内引起严重的并发症。然而,目前尚无批准的用于EV-D68的抗病毒药物或疫苗.在这项研究中,我们发现锌离子在体外对EV-D68感染具有实质性的抗病毒活性。锌盐处理可有效抑制EV-D68RNA复制,蛋白质合成,和感染性病毒体的产生,并在杀病毒浓度(EC50=0.033mM)下抑制细胞病变效应而不产生明显的细胞毒性。氯化锌(ZnCl2)处理适度地抑制EV-D68附着。EV-D68结构蛋白VP1合成的时间剂量分析表明,与细胞裂解物相比,培养基中VP1的抑制作用更强。此外,锌离子载体,吡咯烷二硫代氨基甲酸酯,可以将锌离子输送到细胞中,还增强了ZnCl2处理的抗EV-D68活性。一起来看,我们的结果表明,锌内流的增强可作为EV-D68感染治疗的有效策略.
    Enterovirus D68 (EV-D68) is a respiratory viral pathogen that causes severe respiratory diseases and neurologic manifestations. Since the 2014 outbreak, EV-D68 has been reported to cause severe complications worldwide. However, there are currently no approved antiviral agents or vaccines for EV-D68. In this study, we found that zinc ions exerted substantial antiviral activity against EV-D68 infection in vitro. Zinc salt treatment potently suppressed EV-D68 RNA replication, protein synthesis, and infectious virion production and inhibited cytopathic effects without producing significant cytotoxicity at virucidal concentrations (EC50=0.033mM). Zinc chloride (ZnCl2) treatment moderately inhibited EV-D68 attachment. Time-dose analysis of EV-D68 structural protein VP1 synthesis showed stronger suppression of VP1 in the culture medium than that in the cell lysates. Furthermore, a zinc ionophore, pyrrolidine dithiocarbamate, which can transport zinc ions into cells, also enhanced the anti-EV-D68 activity of ZnCl2 treatment. Taken together, our results demonstrated that the enhancement of zinc influx could serve as a powerful strategy for the therapeutic treatment of EV-D68 infections.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    Human enterovirus D68 (EV-D68) has received considerable attention recently as a global reemergent pathogen because it causes severe respiratory tract infections and acute flaccid myelitis (AFM). The nonstructural protein 2A protease (2Apro) of EVs, which functions in the cleavage of host proteins, comprises a pivotal part of the viral immune evasion process. However, the pathogenic mechanism of EV-D68 is not fully understood. In this study, we found that EV-D68 inhibited antiviral type I interferon responses by cleaving tumor necrosis factor receptor-associated factor 3 (TRAF3), which is the key factor for type I interferon production. EV-D68 inhibited Sendai virus (SEV)-induced interferon regulatory factor 3 (IRF3) activation and beta interferon (IFN-β) expression in HeLa and HEK293T cells. Furthermore, we demonstrated that EV-D68 and 2Apro were able to cleave the C-terminal region of TRAF3 in HeLa and HEK293T cells, respectively. A cysteine-to-alanine substitution at amino acid 107 (C107A) in the 2Apro protease resulted in the loss of cleavage activity to TRAF3, and mutation of glycine at amino acid 462 to alanine (G462A) in TRAF3 conferred resistance to 2Apro These results suggest that control of TRAF3 by 2Apro may be a mechanism EV-D68 utilizes to subvert host innate immune responses.IMPORTANCE Human enterovirus 68 (EV-D68) has received considerable attention recently as a global reemergent pathogen because it causes severe respiratory tract infections and acute flaccid myelitis. The nonstructural protein 2A protease (2Apro) of EV, which functions in cleavage of host proteins, comprises an essential part of the viral immune evasion process. However, the pathogenic mechanism of EV-D68 is not fully understood. Here, we show for the first time that EV-D68 inhibited antiviral type I interferon responses by cleaving tumor necrosis factor receptor-associated factor 3 (TRAF3). Furthermore, we identified the key cleavage site in TRAF3. Our study may suggest a new mechanism by which the 2Apro of EV facilitates subversion of host innate immune responses. These findings increase our understanding of EV-D68 infection and may help identify new antiviral targets against EV-D68.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号